Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Eur Child Adolesc Psychiatry ; 33(2): 431-438, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36847865

RESUMO

Etiologies and the whole picture in childhood mental, behavioral, and neurodevelopmental disorders related to gestational age are unclear. This study included all Finnish children (N = 341,632) born between January 1, 2001, and December 31, 2006, whose data including their mothers (N = 241,284) were collected from national registers. Children with unclear gestational age (GA) (N = 1245), severe congenital malformations (N = 11,746), and moderate/severe/undefined cognitive impairment (N = 1140), and those who died during the perinatal period (N = 599) were excluded. The main outcome was the prevalence of mental and behavioral disorders (International Classification of Disorders) at 0 - 12 years of age in association with GA, adjusted for gender and prenatal variables. Out of all included (N = 326,902) children 16.6% (N = 54,270) were diagnosed to have any mental health disorder at 0 - 12 years. Adjusted Odd Ratio (OR) were for any disorder in preterm (< 37 weeks) 1.37 [1.28 - 1.46] and 4.03 [3.08 - 5.26] in extreme preterm (≤ 28 weeks) versus term born children, p < 0.05. The lower the GA at birth, the higher the risk for multiple disorders and earlier onset of disorder, p < 0.05. Adjusted ORs were for male/female 1.94 [1.90 - 1.99], maternal mental health disorder (yes/not) 1.99 [1.92 - 2.07], and smoking during pregnancy (yes/not) 1.58 [1.54 - 1.62], and these risks were more common in preterm versus term born children (p < 0.05). Extreme early birth was a strong risk factor per se for any or multiple and early shown mental health disorders. Other risk factors for mental health accumulated to preterm children.


Assuntos
Disfunção Cognitiva , Transtornos do Neurodesenvolvimento , Recém-Nascido , Gravidez , Criança , Humanos , Masculino , Feminino , Finlândia/epidemiologia , Transtornos do Neurodesenvolvimento/epidemiologia , Fatores de Risco , Mães
2.
J Biol Chem ; 299(7): 104836, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37209827

RESUMO

Insulin is made from proinsulin, but the extent to which fasting/feeding controls the homeostatically regulated proinsulin pool in pancreatic ß-cells remains largely unknown. Here, we first examined ß-cell lines (INS1E and Min6, which proliferate slowly and are routinely fed fresh medium every 2-3 days) and found that the proinsulin pool size responds to each feeding within 1 to 2 h, affected both by the quantity of fresh nutrients and the frequency with which they are provided. We observed no effect of nutrient feeding on the overall rate of proinsulin turnover as quantified from cycloheximide-chase experiments. We show that nutrient feeding is primarily linked to rapid dephosphorylation of translation initiation factor eIF2α, presaging increased proinsulin levels (and thereafter, insulin levels), followed by its rephosphorylation during the ensuing hours that correspond to a fall in proinsulin levels. The decline of proinsulin levels is blunted by the integrated stress response inhibitor, ISRIB, or by inhibition of eIF2α rephosphorylation with a general control nonderepressible 2 (not PERK) kinase inhibitor. In addition, we demonstrate that amino acids contribute importantly to the proinsulin pool; mass spectrometry shows that ß-cells avidly consume extracellular glutamine, serine, and cysteine. Finally, we show that in both rodent and human pancreatic islets, fresh nutrient availability dynamically increases preproinsulin, which can be quantified without pulse-labeling. Thus, the proinsulin available for insulin biosynthesis is rhythmically controlled by fasting/feeding cycles.


Assuntos
Células Secretoras de Insulina , Nutrientes , Proinsulina , Humanos , Insulina/biossíntese , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Nutrientes/farmacologia , Proinsulina/biossíntese , Proinsulina/metabolismo , Estresse Fisiológico , Transdução de Sinais , Linhagem Celular , Regulação para Cima
3.
Nat Commun ; 12(1): 5991, 2021 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-34645803

RESUMO

The reticulon-3 (RTN3)-driven targeting complex promotes clearance of misfolded prohormones from the endoplasmic reticulum (ER) for lysosomal destruction by ER-phagy. Because RTN3 resides in the cytosolic leaflet of the ER bilayer, the mechanism of selecting misfolded prohormones as ER-phagy cargo on the luminal side of the ER membrane remains unknown. Here we identify the ER transmembrane protein PGRMC1 as an RTN3-binding partner. Via its luminal domain, PGRMC1 captures misfolded prohormones, targeting them for RTN3-dependent ER-phagy. PGRMC1 selects cargos that are smaller than the large size of other reported ER-phagy substrates. Cargos for PGRMC1 include mutant proinsulins that block secretion of wildtype proinsulin through dominant-negative interactions within the ER, causing insulin-deficiency. Chemical perturbation of PGRMC1 partially restores WT insulin storage by preventing ER-phagic degradation of WT and mutant proinsulin. Thus, PGRMC1 acts as a size-selective cargo receptor during RTN3-dependent ER-phagy, and is a potential therapeutic target for diabetes.


Assuntos
Proteínas de Transporte/genética , Retículo Endoplasmático/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Proinsulina/genética , Receptores de Progesterona/genética , Animais , Autofagia/genética , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Retículo Endoplasmático/genética , Expressão Gênica , Células HEK293 , Humanos , Células Secretoras de Insulina/citologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Lisossomos/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Mutação , Proteínas do Tecido Nervoso/metabolismo , Cultura Primária de Células , Proinsulina/metabolismo , Ligação Proteica , Domínios Proteicos , Dobramento de Proteína , Proteólise , Ratos , Receptores de Progesterona/metabolismo
4.
Cell Mol Life Sci ; 78(16): 6017-6031, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34245311

RESUMO

A precondition for efficient proinsulin export from the endoplasmic reticulum (ER) is that proinsulin meets ER quality control folding requirements, including formation of the Cys(B19)-Cys(A20) "interchain" disulfide bond, facilitating formation of the Cys(B7)-Cys(A7) bridge. The third proinsulin disulfide, Cys(A6)-Cys(A11), is not required for anterograde trafficking, i.e., a "lose-A6/A11" mutant [Cys(A6), Cys(A11) both converted to Ser] is well secreted. Nevertheless, an unpaired Cys(A11) can participate in disulfide mispairings, causing ER retention of proinsulin. Among the many missense mutations causing the syndrome of Mutant INS gene-induced Diabetes of Youth (MIDY), all seem to exhibit perturbed proinsulin disulfide bond formation. Here, we have examined a series of seven MIDY mutants [including G(B8)V, Y(B26)C, L(A16)P, H(B5)D, V(B18)A, R(Cpep + 2)C, E(A4)K], six of which are essentially completely blocked in export from the ER in pancreatic ß-cells. Three of these mutants, however, must disrupt the Cys(A6)-Cys(A11) pairing to expose a critical unpaired cysteine thiol perturbation of proinsulin folding and ER export, because when introduced into the proinsulin lose-A6/A11 background, these mutants exhibit native-like disulfide bonding and improved trafficking. This maneuver also ameliorates dominant-negative blockade of export of co-expressed wild-type proinsulin. A growing molecular understanding of proinsulin misfolding may permit allele-specific pharmacological targeting for some MIDY mutants.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Proinsulina/metabolismo , Adolescente , Células Cultivadas , Cisteína/genética , Cisteína/metabolismo , Diabetes Mellitus Tipo 2/genética , Dissulfetos/metabolismo , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Humanos , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Mutação de Sentido Incorreto/genética , Proinsulina/genética , Dobramento de Proteína
5.
JCI Insight ; 6(5)2021 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-33529174

RESUMO

Limitations in cell proliferation are important for normal function of differentiated tissues and essential for the safety of cell replacement products made from pluripotent stem cells, which have unlimited proliferative potential. To evaluate whether these limitations can be established pharmacologically, we exposed pancreatic progenitors differentiating from human pluripotent stem cells to small molecules that interfere with cell cycle progression either by inducing G1 arrest or by impairing S phase entry or S phase completion and determined growth potential, differentiation, and function of insulin-producing endocrine cells. We found that the combination of G1 arrest with a compromised ability to complete DNA replication promoted the differentiation of pancreatic progenitor cells toward insulin-producing cells and could substitute for endocrine differentiation factors. Reduced replication fork speed during differentiation improved the stability of insulin expression, and the resulting cells protected mice from diabetes without the formation of cystic growths. The proliferative potential of grafts was proportional to the reduction of replication fork speed during pancreatic differentiation. Therefore, a compromised ability to enter and complete S phase is a functionally important property of pancreatic endocrine differentiation, can be achieved by reducing replication fork speed, and is an important determinant of cell-intrinsic limitations of growth.


Assuntos
Ciclo Celular , Diferenciação Celular , Replicação do DNA , Diabetes Mellitus , Células-Tronco Pluripotentes Induzidas , Células Secretoras de Insulina , Transplante de Células-Tronco , Animais , Afidicolina , Proliferação de Células , Diabetes Mellitus/terapia , Humanos , Insulina/metabolismo , Ilhotas Pancreáticas , Camundongos , Pâncreas , Células-Tronco Pluripotentes , Transplantes
6.
Proc Natl Acad Sci U S A ; 117(47): 29618-29628, 2020 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-33154160

RESUMO

Proteins have evolved to be foldable, and yet determinants of foldability may be inapparent once the native state is reached. Insight has emerged from studies of diseases of protein misfolding, exemplified by monogenic diabetes mellitus due to mutations in proinsulin leading to endoplasmic reticulum stress and ß-cell death. Cellular foldability of human proinsulin requires an invariant Phe within a conserved crevice at the receptor-binding surface (position B24). Any substitution, even related aromatic residue TyrB24, impairs insulin biosynthesis and secretion. As a seeming paradox, a monomeric TyrB24 insulin analog exhibits a native-like structure in solution with only a modest decrement in stability. Packing of TyrB24 is similar to that of PheB24, adjoining core cystine B19-A20 to seal the core; the analog also exhibits native self-assembly. Although affinity for the insulin receptor is decreased ∼20-fold, biological activities in cells and rats were within the range of natural variation. Together, our findings suggest that the invariance of PheB24 among vertebrate insulins and insulin-like growth factors reflects an essential role in enabling efficient protein folding, trafficking, and secretion, a function that is inapparent in native structures. In particular, we envision that the para-hydroxyl group of TyrB24 hinders pairing of cystine B19-A20 in an obligatory on-pathway folding intermediate. The absence of genetic variation at B24 and other conserved sites near this disulfide bridge-excluded due to ß-cell dysfunction-suggests that insulin has evolved to the edge of foldability. Nonrobustness of a protein's fitness landscape underlies both a rare monogenic syndrome and "diabesity" as a pandemic disease of civilization.


Assuntos
Insulina/metabolismo , Substituição de Aminoácidos/fisiologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Diabetes Mellitus/metabolismo , Dissulfetos/metabolismo , Redes Reguladoras de Genes/fisiologia , Células HEK293 , Humanos , Células Secretoras de Insulina/metabolismo , Células MCF-7 , Proinsulina/metabolismo , Ligação Proteica/fisiologia , Dobramento de Proteína , Ratos , Receptor de Insulina/metabolismo , Relação Estrutura-Atividade
7.
Mol Metab ; 40: 101015, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32416313

RESUMO

OBJECTIVE: Risk alleles for type 2 diabetes at the STARD10 locus are associated with lowered STARD10 expression in the ß-cell, impaired glucose-induced insulin secretion, and decreased circulating proinsulin:insulin ratios. Although likely to serve as a mediator of intracellular lipid transfer, the identity of the transported lipids and thus the pathways through which STARD10 regulates ß-cell function are not understood. The aim of this study was to identify the lipids transported and affected by STARD10 in the ß-cell and the role of the protein in controlling proinsulin processing and insulin granule biogenesis and maturation. METHODS: We used isolated islets from mice deleted selectively in the ß-cell for Stard10 (ßStard10KO) and performed electron microscopy, pulse-chase, RNA sequencing, and lipidomic analyses. Proteomic analysis of STARD10 binding partners was executed in the INS1 (832/13) cell line. X-ray crystallography followed by molecular docking and lipid overlay assay was performed on purified STARD10 protein. RESULTS: ßStard10KO islets had a sharply altered dense core granule appearance, with a dramatic increase in the number of "rod-like" dense cores. Correspondingly, basal secretion of proinsulin was increased versus wild-type islets. The solution of the crystal structure of STARD10 to 2.3 Å resolution revealed a binding pocket capable of accommodating polyphosphoinositides, and STARD10 was shown to bind to inositides phosphorylated at the 3' position. Lipidomic analysis of ßStard10KO islets demonstrated changes in phosphatidylinositol levels, and the inositol lipid kinase PIP4K2C was identified as a STARD10 binding partner. Also consistent with roles for STARD10 in phosphoinositide signalling, the phosphoinositide-binding proteins Pirt and Synaptotagmin 1 were amongst the differentially expressed genes in ßStard10KO islets. CONCLUSION: Our data indicate that STARD10 binds to, and may transport, phosphatidylinositides, influencing membrane lipid composition, insulin granule biosynthesis, and insulin processing.


Assuntos
Diabetes Mellitus Tipo 2/genética , Fosfoproteínas/metabolismo , Alelos , Animais , Proteínas de Transporte/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animais de Doenças , Feminino , Insulina/metabolismo , Secreção de Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Metabolismo dos Lipídeos/genética , Metabolismo dos Lipídeos/fisiologia , Lipídeos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Simulação de Acoplamento Molecular , Fosfatidilinositóis/metabolismo , Fosfoproteínas/genética , Ligação Proteica , Proteômica , Fatores de Risco , Vesículas Secretórias/metabolismo
8.
Elife ; 82019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31184302

RESUMO

Biosynthesis of insulin - critical to metabolic homeostasis - begins with folding of the proinsulin precursor, including formation of three evolutionarily conserved intramolecular disulfide bonds. Remarkably, normal pancreatic islets contain a subset of proinsulin molecules bearing at least one free cysteine thiol. In human (or rodent) islets with a perturbed endoplasmic reticulum folding environment, non-native proinsulin enters intermolecular disulfide-linked complexes. In genetically obese mice with otherwise wild-type islets, disulfide-linked complexes of proinsulin are more abundant, and leptin receptor-deficient mice, the further increase of such complexes tracks with the onset of islet insulin deficiency and diabetes. Proinsulin-Cys(B19) and Cys(A20) are necessary and sufficient for the formation of proinsulin disulfide-linked complexes; indeed, proinsulin Cys(B19)-Cys(B19) covalent homodimers resist reductive dissociation, highlighting a structural basis for aberrant proinsulin complex formation. We conclude that increased proinsulin misfolding via disulfide-linked complexes is an early event associated with prediabetes that worsens with ß-cell dysfunction in type two diabetes.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Proinsulina/química , Dobramento de Proteína , Animais , Cisteína/química , Cisteína/genética , Cisteína/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Progressão da Doença , Dissulfetos/química , Dissulfetos/metabolismo , Retículo Endoplasmático/metabolismo , Chaperona BiP do Retículo Endoplasmático , Humanos , Ilhotas Pancreáticas/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Proinsulina/genética , Proinsulina/metabolismo , Receptores para Leptina/deficiência , Receptores para Leptina/genética
9.
Diabetes ; 68(4): 747-760, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30670477

RESUMO

Although endoplasmic reticulum (ER) chaperone binding to mutant proinsulin has been reported, the role of protein chaperones in the handling of wild-type proinsulin is underinvestigated. Here, we have explored the importance of glucose-regulated protein 94 (GRP94), a prominent ER chaperone known to fold insulin-like growth factors, in proinsulin handling within ß-cells. We found that GRP94 coimmunoprecipitated with proinsulin and that inhibition of GRP94 function and/or expression reduced glucose-dependent insulin secretion, shortened proinsulin half-life, and lowered intracellular proinsulin and insulin levels. This phenotype was accompanied by post-ER proinsulin misprocessing and higher numbers of enlarged insulin granules that contained amorphic material with reduced immunogold staining for mature insulin. Insulin granule exocytosis was accelerated twofold, but the secreted insulin had diminished bioactivity. Moreover, GRP94 knockdown or knockout in ß-cells selectively activated protein kinase R-like endoplasmic reticulum kinase (PERK), without increasing apoptosis levels. Finally, GRP94 mRNA was overexpressed in islets from patients with type 2 diabetes. We conclude that GRP94 is a chaperone crucial for proinsulin handling and insulin secretion.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Secreção de Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Proteínas de Membrana/metabolismo , Proinsulina/metabolismo , Animais , Apoptose/fisiologia , Linhagem Celular Tumoral , Estresse do Retículo Endoplasmático/fisiologia , Exocitose/fisiologia , Humanos , Insulina/metabolismo , Dobramento de Proteína , Ratos , eIF-2 Quinase/metabolismo
10.
Diabetes Obes Metab ; 20 Suppl 2: 28-50, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30230185

RESUMO

Insulin synthesis in pancreatic ß-cells is initiated as preproinsulin. Prevailing glucose concentrations, which oscillate pre- and postprandially, exert major dynamic variation in preproinsulin biosynthesis. Accompanying upregulated translation of the insulin precursor includes elements of the endoplasmic reticulum (ER) translocation apparatus linked to successful orientation of the signal peptide, translocation and signal peptide cleavage of preproinsulin-all of which are necessary to initiate the pathway of proper proinsulin folding. Evolutionary pressures on the primary structure of proinsulin itself have preserved the efficiency of folding ("foldability"), and remarkably, these evolutionary pressures are distinct from those protecting the ultimate biological activity of insulin. Proinsulin foldability is manifest in the ER, in which the local environment is designed to assist in the overall load of proinsulin folding and to favour its disulphide bond formation (while limiting misfolding), all of which is closely tuned to ER stress response pathways that have complex (beneficial, as well as potentially damaging) effects on pancreatic ß-cells. Proinsulin misfolding may occur as a consequence of exuberant proinsulin biosynthetic load in the ER, proinsulin coding sequence mutations, or genetic predispositions that lead to an altered ER folding environment. Proinsulin misfolding is a phenotype that is very much linked to deficient insulin production and diabetes, as is seen in a variety of contexts: rodent models bearing proinsulin-misfolding mutants, human patients with Mutant INS-gene-induced Diabetes of Youth (MIDY), animal models and human patients bearing mutations in critical ER resident proteins, and, quite possibly, in more common variety type 2 diabetes.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/biossíntese , Precursores de Proteínas/biossíntese , Animais , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Modelos Animais de Doenças , Retículo Endoplasmático/metabolismo , Humanos , Insulina/química , Camundongos , Mutação/genética , Proinsulina/biossíntese , Proinsulina/química , Proinsulina/genética , Dobramento de Proteína , Precursores de Proteínas/química , Sistemas de Translocação de Proteínas/metabolismo
12.
J Clin Invest ; 128(3): 1125-1140, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29457782

RESUMO

Pro-opiomelanocortin (POMC) neurons function as key regulators of metabolism and physiology by releasing prohormone-derived neuropeptides with distinct biological activities. However, our understanding of early events in prohormone maturation in the ER remains incomplete. Highlighting the significance of this gap in knowledge, a single POMC cysteine-to-phenylalanine mutation at position 28 (POMC-C28F) is defective for ER processing and causes early onset obesity in a dominant-negative manner in humans through an unclear mechanism. Here, we report a pathologically important role of Sel1L-Hrd1, the protein complex of ER-associated degradation (ERAD), within POMC neurons. Mice with POMC neuron-specific Sel1L deficiency developed age-associated obesity due, at least in part, to the ER retention of POMC that led to hyperphagia. The Sel1L-Hrd1 complex targets a fraction of nascent POMC molecules for ubiquitination and proteasomal degradation, preventing accumulation of misfolded and aggregated POMC, thereby ensuring that another fraction of POMC can undergo normal posttranslational processing and trafficking for secretion. Moreover, we found that the disease-associated POMC-C28F mutant evades ERAD and becomes aggregated due to the presence of a highly reactive unpaired cysteine thiol at position 50. Thus, this study not only identifies ERAD as an important mechanism regulating POMC maturation within the ER, but also provides insights into the pathogenesis of monogenic obesity associated with defective prohormone folding.


Assuntos
Degradação Associada com o Retículo Endoplasmático , Retículo Endoplasmático/patologia , Hipotálamo/patologia , Obesidade/patologia , Pró-Opiomelanocortina/metabolismo , Animais , Axônios , Cisteína/química , Comportamento Alimentar , Feminino , Proteínas de Fluorescência Verde/metabolismo , Humanos , Inflamação , Peptídeos e Proteínas de Sinalização Intracelular , Leptina/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Neurônios/metabolismo , Fenilalanina/química , Pró-Opiomelanocortina/genética , Proteínas/metabolismo , Compostos de Sulfidrila , Ubiquitina/química , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
13.
Cell Metab ; 26(3): 568-575.e3, 2017 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-28877460

RESUMO

The canonical notion that type 1 diabetes (T1D) results following a complete destruction of ß cells has recently been questioned as small amounts of C-peptide are detectable in patients with long-standing disease. We analyzed protein and gene expression levels for proinsulin, insulin, C-peptide, and islet amyloid polypeptide within pancreatic tissues from T1D, autoantibody positive (Ab+), and control organs. Insulin and C-peptide levels were low to undetectable in extracts from the T1D cohort; however, proinsulin and INS mRNA were detected in the majority of T1D pancreata. Interestingly, heterogeneous nuclear RNA (hnRNA) for insulin and INS-IGF2, both originating from the INS promoter, were essentially undetectable in T1D pancreata, arguing for a silent INS promoter. Expression of PCSK1, a convertase responsible for proinsulin processing, was reduced in T1D pancreata, supportive of persistent proinsulin. These data implicate the existence of ß cells enriched for inefficient insulin/C-peptide production in T1D patients, potentially less susceptible to autoimmune destruction.


Assuntos
Diabetes Mellitus Tipo 1/genética , Insulina/genética , Pâncreas/metabolismo , Proinsulina/metabolismo , Adulto , Diabetes Mellitus Tipo 1/patologia , Feminino , Regulação da Expressão Gênica , Glucagon/metabolismo , Humanos , Insulina/metabolismo , Masculino , Pâncreas/patologia , Pâncreas Exócrino/metabolismo , Pâncreas Exócrino/patologia , Pró-Proteína Convertase 1/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Adulto Jovem
14.
Brain Behav ; 6(2): e00431, 2016 02.
Artigo em Inglês | MEDLINE | ID: mdl-26811802

RESUMO

OBJECTIVES: This study aimed at ascertaining the standardized mortality ratios (SMR) for those with an intellectual disability (ID) in Finland. MATERIALS AND METHODS: We used the statistical database of the national insurance institution of Finland and Statistics Finland's mean population figures. We determined the number of individuals who received benefits (disability allowance, disability pension, or care allowance for pensioners) due to an ID diagnosis and the number of those whose benefit had been terminated due to death during the years 1996-2011. RESULTS: SMR for females with a mild ID (IQ 50-69) was 2.8 (95% CI: 2.60-3.01) and for males 2.0 (95% CI: 1.88-2.14), and for females with a severe ID (IQ <50) 5.2 (95% CI: 4.99-5.50) and for males 2.6 (95% CI: 2.48-2.72). CONCLUSION: This significant difference in the SMR figures between males and females with ID warrants further research.


Assuntos
Deficiência Intelectual/mortalidade , Feminino , Finlândia/epidemiologia , Humanos , Masculino , Programas Nacionais de Saúde , Índice de Gravidade de Doença
15.
Cell Rep ; 13(11): 2527-2538, 2015 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-26673325

RESUMO

Nutrient levels dictate the activity of O-linked N-acetylglucosamine transferase (OGT) to regulate O-GlcNAcylation, a post-translational modification mechanism to "fine-tune" intracellular signaling and metabolic status. However, the requirement of O-GlcNAcylation for maintaining glucose homeostasis by regulating pancreatic ß cell mass and function is unclear. Here, we reveal that mice lacking ß cell OGT (ßOGT-KO) develop diabetes and ß cell failure. ßOGT-KO mice demonstrated increased ER stress and distended ER architecture, and these changes ultimately caused the loss of ß cell mass due to ER-stress-induced apoptosis and decreased proliferation. Akt1/2 signaling was also dampened in ßOGT-KO islets. The mechanistic role of these processes was demonstrated by rescuing the phenotype of ßOGT-KO mice with concomitant Chop gene deletion or genetic reconstitution of Akt2. These findings identify OGT as a regulator of ß cell mass and function and provide a direct link between O-GlcNAcylation and ß cell survival by regulation of ER stress responses and modulation of Akt1/2 signaling.


Assuntos
Acetilglucosamina/metabolismo , Estresse do Retículo Endoplasmático , Células Secretoras de Insulina/metabolismo , N-Acetilglucosaminiltransferases/genética , Envelhecimento , Animais , Apoptose , Proliferação de Células , Regulação para Baixo , Feminino , Teste de Tolerância a Glucose , Hiperglicemia/etiologia , Hiperglicemia/metabolismo , Hiperglicemia/veterinária , Insulina/metabolismo , Secreção de Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , N-Acetilglucosaminiltransferases/deficiência , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo
16.
EMBO J ; 34(13): 1759-72, 2015 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-25908839

RESUMO

Directed differentiation of human pluripotent stem cells into functional insulin-producing beta-like cells holds great promise for cell replacement therapy for patients suffering from diabetes. This approach also offers the unique opportunity to study otherwise inaccessible aspects of human beta cell development and function in vitro. Here, we show that current pancreatic progenitor differentiation protocols promote precocious endocrine commitment, ultimately resulting in the generation of non-functional polyhormonal cells. Omission of commonly used BMP inhibitors during pancreatic specification prevents precocious endocrine formation while treatment with retinoic acid followed by combined EGF/KGF efficiently generates both PDX1(+) and subsequent PDX1(+)/NKX6.1(+) pancreatic progenitor populations, respectively. Precise temporal activation of endocrine differentiation in PDX1(+)/NKX6.1(+) progenitors produces glucose-responsive beta-like cells in vitro that exhibit key features of bona fide human beta cells, remain functional after short-term transplantation, and reduce blood glucose levels in diabetic mice. Thus, our simplified and scalable system accurately recapitulates key steps of human pancreas development and provides a fast and reproducible supply of functional human beta-like cells.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células-Tronco Embrionárias/fisiologia , Células Secretoras de Insulina/fisiologia , Pâncreas/citologia , Animais , Glicemia/metabolismo , Células Cultivadas , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/terapia , Células-Tronco Embrionárias/citologia , Glucose/farmacologia , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/transplante , Camundongos , Camundongos SCID , Camundongos Transgênicos , Estreptozocina
17.
J Biol Chem ; 288(3): 1896-906, 2013 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-23223446

RESUMO

Classically, exit from the endoplasmic reticulum (ER) is rate-limiting for secretory protein trafficking because protein folding/assembly occurs there. In this study, we have exploited "hPro-CpepSfGFP," a human proinsulin bearing "superfolder" green fluorescent C-peptide expressed in pancreatic ß cells where it is processed to human insulin and CpepSfGFP. Remarkably, steady-state accumulation of hPro-CpepSfGFP and endogenous proinsulin is in the Golgi region, as if final stages of protein folding/assembly were occurring there. The Golgi regional distribution of proinsulin is dynamic, influenced by fasting/refeeding, and increased with ß cell zinc deficiency. However, coexpression of ER-entrapped mutant proinsulin-C(A7)Y shifts the steady-state distribution of wild-type proinsulin to the ER. Endogenous proinsulin coprecipitates with hPro-CpepSfGFP and even more so with hProC(A7)Y-CpepSfGFP. Using Cerulean and Venus-tagged proinsulins, we find that both WT-WT and WT-mutant proinsulin pairs exhibit FRET. The data demonstrate that wild-type proinsulin dimerizes within the ER but accumulates at a poorly recognized slow step within the Golgi region, reflecting either slow kinetics of proinsulin hexamerization, steps in formation of nascent secretory granules, or other unknown molecular events. However, in the presence of ongoing misfolding of a subpopulation of proinsulin in ß cells, the rate-limiting step in transport of the remaining proinsulin shifts to the ER.


Assuntos
Peptídeo C/metabolismo , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animais , Peptídeo C/química , Peptídeo C/genética , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Dimerização , Retículo Endoplasmático/genética , Retículo Endoplasmático/ultraestrutura , Expressão Gênica , Complexo de Golgi/genética , Complexo de Golgi/ultraestrutura , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Insulina/química , Insulina/genética , Células Secretoras de Insulina/citologia , Cinética , Camundongos , Microscopia Confocal , Plasmídeos , Ligação Proteica , Dobramento de Proteína , Transporte Proteico , Ratos , Transfecção
18.
BMC Biochem ; 11: 48, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21122136

RESUMO

BACKGROUND: RhoGDI proteins are important regulators of the small GTPase Rac, because they shuttle Rac from the cytoplasm to membranes and also protect Rac from activation, deactivation and degradation. How the binding and release of Rac from RhoGDI is regulated is not precisely understood. RESULTS: We report that the non-receptor tyrosine kinase Fer is able to phosphorylate RhoGDIα and form a direct protein complex with it. This interaction is mediated by the C-terminal end of RhoGDIα. Activation of Fer by reactive oxygen species caused increased phosphorylation of RhoGDIα and pervanadate treatment further augmented this. Tyrosine phosphorylation of RhoGDIα by Fer prevented subsequent binding of Rac to RhoGDIα, but once a RhoGDIα-Rac complex was formed, the Fer kinase was not able to cause Rac release through tyrosine phosphorylation of preformed RhoGDIα-Rac complexes. CONCLUSIONS: These results identify tyrosine phosphorylation of RhoGDIα by Fer as a mechanism to regulate binding of RhoGDIα to Rac.


Assuntos
Inibidores de Dissociação do Nucleotídeo Guanina/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Inibidores de Dissociação do Nucleotídeo Guanina/química , Humanos , Fosforilação , Ligação Proteica , Ratos , Especificidade por Substrato , Tirosina/metabolismo , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho , Inibidores da Dissociação do Nucleotídeo Guanina rho-Específico
19.
PLoS One ; 5(10): e13333, 2010 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-20948967

RESUMO

Recently, a syndrome of Mutant INS-gene-induced Diabetes of Youth (MIDY, derived from one of 26 distinct mutations) has been identified as a cause of insulin-deficient diabetes, resulting from expression of a misfolded mutant proinsulin protein in the endoplasmic reticulum (ER) of insulin-producing pancreatic beta cells. Genetic deletion of one, two, or even three alleles encoding insulin in mice does not necessarily lead to diabetes. Yet MIDY patients are INS-gene heterozygotes; inheritance of even one MIDY allele, causes diabetes. Although a favored explanation for the onset of diabetes is that insurmountable ER stress and ER stress response from the mutant proinsulin causes a net loss of beta cells, in this report we present three surprising and interlinked discoveries. First, in the presence of MIDY mutants, an increased fraction of wild-type proinsulin becomes recruited into nonnative disulfide-linked protein complexes. Second, regardless of whether MIDY mutations result in the loss, or creation, of an extra unpaired cysteine within proinsulin, Cys residues in the mutant protein are nevertheless essential in causing intracellular entrapment of co-expressed wild-type proinsulin, blocking insulin production. Third, while each of the MIDY mutants induces ER stress and ER stress response; ER stress and ER stress response alone appear insufficient to account for blockade of wild-type proinsulin. While there is general agreement that ultimately, as diabetes progresses, a significant loss of beta cell mass occurs, the early events described herein precede cell death and loss of beta cell mass. We conclude that the molecular pathogenesis of MIDY is initiated by perturbation of the disulfide-coupled folding pathway of wild-type proinsulin.


Assuntos
Mutação , Proinsulina/genética , Proinsulina/metabolismo , Sequência de Aminoácidos , Transporte Biológico , Retículo Endoplasmático/metabolismo , Genes Dominantes , Humanos , Dados de Sequência Molecular , Proinsulina/química
20.
J Pediatr ; 156(2): 185-90.e1, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19818449

RESUMO

OBJECTIVE: To evaluate the association between maternal smoking during pregnancy and both brain volumes and head circumference in very-low-birth-weight/very-low-gestational-age infants. STUDY DESIGN: The PIPARI Study is a prospective follow-up study of infants with a birth weight < or =1500 g or a gestational age <32 weeks born in 2001 to 2006 (n = 232) at Turku University Hospital. The brain was imaged by serial brain ultrasound examinations until discharge and magnetic resonance imaging at term age. The head circumference was measured at birth, term, and 2 years corrected age. These measures were correlated to maternal smoking during pregnancy as reported by the mothers. RESULTS: The prevalence of maternal smoking was 18%. The frontal lobe (P = .01) and the cerebellar (P = .03) volumes were significantly smaller in the exposed than in the unexposed infants. The volumes of the other parts of the brain did not differ. There was no association between prenatal smoking exposure and head growth or structural brain disease. CONCLUSIONS: Prenatal smoking exposure was associated with significantly smaller frontal lobe and cerebellar volumes in the brains of preterm infants. This is consistent with reports showing an association between prenatal smoking exposure and impairments in frontal lobe and cerebellar functions such as emotion, impulse control, and attention.


Assuntos
Encéfalo/patologia , Recém-Nascido Prematuro , Recém-Nascido de muito Baixo Peso , Fumar/efeitos adversos , Encéfalo/embriologia , Cerebelo/patologia , Ecoencefalografia , Feminino , Desenvolvimento Fetal , Finlândia/epidemiologia , Lobo Frontal/patologia , Humanos , Recém-Nascido , Imageamento por Ressonância Magnética , Masculino , Tamanho do Órgão , Gravidez , Prevalência , Estudos Prospectivos , Fumar/epidemiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA